Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Targeting RAS signalling pathways in cancer therapy

Key Points

  • RAS proteins, their regulators and the downstream enzymes that they control are activated in many tumour types by a variety of mechanisms, including oncogenic mutation of RAS genes.

  • They are crucial mediators of several of the malignant characteristics of transformed cells and are therefore good candidates for tumour therapy.

  • RAS proteins require post-translational modification by farnesylation to be biologically active. Farnesyl transferase inhibitors have some antitumour activity in the clinic, but they seem to act through targets other than RAS.

  • Kinase inhibitors that block either RAF or mitogen-activated protein (MAP) kinase kinase MEK in the RAF/MAP kinase pathway downstream of RAS have been developed and show promise in early clinical trials.

  • Inhibitors acting on epidermal growth factor (EGF) receptor and ERBB2 upstream activators of RAS have been developed. Antibodies directed against ERBB2 have been licensed for the treatment of breast cancer, whereas small-molecule EGF receptor inhibitors show potential against lung cancer in clinical trials.

  • Other RAS-related therapies are in development, including inhibitors of AKT/PKB kinase activity, which is activated by RAS oncogenic mutation and by PTEN tumour-suppressor gene loss.

Abstract

The RAS proteins control signalling pathways that are key regulators of several aspects of normal cell growth and malignant transformation. They are aberrant in most human tumours due to activating mutations in the RAS genes themselves or to alterations in upstream or downstream signalling components. Rational therapies that target the RAS pathways might inhibit tumour growth, survival and spread. Several of these new therapeutic agents are showing promise in the clinic and many more are being developed.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Signalling upstream of RAS.
Figure 2: Signalling downstream of RAS.

Similar content being viewed by others

References

  1. Barbacid, M. Ras genes. Annu. Rev. Biochem. 56, 779–827 (1987).

    Article  CAS  PubMed  Google Scholar 

  2. Bos, J. L. Ras oncogenes in human cancer: a review. Cancer Res. 49, 4682–4689 (1989).

    CAS  PubMed  Google Scholar 

  3. Shields, J. M., Pruitt, K., McFall, A., Shaub, A. & Der, C. J. Understanding Ras: 'it ain't over 'til it's over'. Trends Cell Biol. 10, 147–154 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Campbell, S. L., Khosravi-Far, R., Rossman, K. L., Clark, G. J. & Der, C. J. Increasing complexity of Ras signaling. Oncogene 17, 1395–1413 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Wittinghofer, A. Signal transduction via Ras. Biol. Chem. 379, 933–937 (1998).

    CAS  PubMed  Google Scholar 

  6. Lowy, D. R. & Willumsen, B. M. Function and regulation of ras. Annu. Rev. Biochem. 62, 851–891 (1993).

    Article  CAS  PubMed  Google Scholar 

  7. Johnson, L. et al. K-ras is an essential gene in the mouse with partial functional overlap with N-ras. Genes Dev. 11, 2468–2481 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Seabra, M. C. Membrane association and targeting of prenylated Ras-like GTPases. Cell Signal. 10, 167–172 (1998).

    Article  CAS  PubMed  Google Scholar 

  9. Cox, A. D. & Der, C. J. Farnesyltransferase inhibitors and cancer treatment: targeting simply Ras? Biochim. Biophys. Acta 1333, F51–F71 (1997).

    CAS  PubMed  Google Scholar 

  10. Hancock, J. F., Magee, A. I., Childs, J. E. & Marshall, C. J. All ras proteins are polyisoprenylated but only some are palmitoylated. Cell 57, 1167–1177 (1989).

    Article  CAS  PubMed  Google Scholar 

  11. Hancock, J. F., Paterson, H. & Marshall, C. J. A polybasic domain or palmitoylation is required in addition to the CAAX motif to localize p21ras to the plasma membrane. Cell 63, 133–139 (1990). References 10 and 11 describe the original characterization of the modification of RAS by farnesylation and, in the case of HRAS and NRAS, but not KRAS, palmitoylation.

    Article  CAS  PubMed  Google Scholar 

  12. Reuther, G. W. & Der, C. J. The Ras branch of small GTPases: Ras family members don't fall far from the tree. Curr. Opin. Cell Biol. 12, 157–165 (2000).

    Article  CAS  PubMed  Google Scholar 

  13. Takai, Y., Sasaki, T. & Matozaki, T. Small GTP-binding proteins. Physiol. Rev. 81, 153–208 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Cullen, P. J. & Lockyer, P. J. Integration of calcium and Ras signalling. Nature Rev. Mol. Cell Biol. 3, 339–348 (2002).

    Article  CAS  Google Scholar 

  15. Daub, H., Weiss, F. U., Wallasch, C. & Ullrich, A. Role of transactivation of the EGF receptor in signalling by G-protein-coupled receptors. Nature 379, 557–560 (1996).

    Article  CAS  PubMed  Google Scholar 

  16. Chiu, V. K. et al. Ras signalling on the endoplasmic reticulum and the Golgi. Nature Cell Biol. 4, 343–350 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Ebinu, J. O. et al. RasGRP, a Ras guanyl nucleotide-releasing protein with calcium- and diacylglycerol-binding motifs. Science 280, 1082–1086 (1998).

    Article  CAS  PubMed  Google Scholar 

  18. Downward, J., Graves, J. D., Warne, P. H., Rayter, S. & Cantrell, D. A. Stimulation of p21ras upon T-cell activation. Nature 346, 719–723 (1990).

    Article  CAS  PubMed  Google Scholar 

  19. Donovan, S., Shannon, K. M. & Bollag, G. GTPase activating proteins: critical regulators of intracellular signaling. Biochim. Biophys. Acta 1602, 23–45 (2002).

    CAS  PubMed  Google Scholar 

  20. Leevers, S. J., Paterson, H. F. & Marshall, C. J. Requirement for Ras in Raf activation is overcome by targeting Raf to the plasma membrane. Nature 369, 411–414 (1994).

    Article  CAS  PubMed  Google Scholar 

  21. Marais, R., Light, Y., Paterson, H. F. & Marshall, C. J. Ras recruits Raf-1 to the plasma membrane for activation by tyrosine phosphorylation. EMBO J. 14, 3136–3145 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yordy, J. S. & Muise-Helmericks, R. C. Signal transduction and the Ets family of transcription factors. Oncogene 19, 6503–6513 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Pruitt, K. & Der, C. J. Ras and Rho regulation of the cell cycle and oncogenesis. Cancer Lett. 171, 1–10 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Rodriguez-Viciana, P. et al. Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature 370, 527–532 (1994).

    Article  CAS  PubMed  Google Scholar 

  25. Pacold, M. E. et al. Crystal structure and functional analysis of Ras binding to its effector phosphoinositide 3-kinaseγ. Cell 103, 931–943 (2000). This describes the three-dimensional structure of RAS in complex with one of its effectors, PI3K. The size of the interaction interface shows the difficulty that is likely to face the design of inhibitors of protein–protein interaction.

    Article  CAS  PubMed  Google Scholar 

  26. Datta, S. R., Brunet, A. & Greenberg, M. E. Cellular survival: a play in three Akts. Genes Dev. 13, 2905–2927 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Khwaja, A., Rodriguez-Viciana, P., Wennstrom, S., Warne, P. H. & Downward, J. Matrix adhesion and Ras transformation both activate a phosphoinositide 3-OH kinase and protein kinase B/Akt cellular survival pathway. EMBO J. 16, 2783–2793 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lambert, J. M. et al. Tiam1 mediates Ras activation of Rac by a PI(3)K-independent mechanism. Nature Cell Biol. 4, 621–625 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Malliri, A. et al. Mice deficient in the Rac activator Tiam1 are resistant to Ras-induced skin tumours. Nature 417, 867–871 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. De Ruiter, N. D., Burgering, B. M. & Bos, J. L. Regulation of the Forkhead transcription factor AFX by Ral-dependent phosphorylation of threonines 447 and 451. Mol. Cell. Biol. 21, 8225–8235 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kelley, G. G., Reks, S. E., Ondrako, J. M. & Smrcka, A. V. Phospholipase Cε: a novel Ras effector. EMBO J. 20, 743–754 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Weiss, B., Bollag, G. & Shannon, K. Hyperactive Ras as a therapeutic target in neurofibromatosis type 1. Am. J. Med. Genet. 89, 14–22 (1999).

    Article  CAS  PubMed  Google Scholar 

  33. Mendelsohn, J. & Baselga, J. The EGF receptor family as targets for cancer therapy. Oncogene 19, 6550–6565 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. Kuan, C. T., Wikstrand, C. J. & Bigner, D. D. EGF mutant receptor vIII as a molecular target in cancer therapy. Endocr. Relat. Cancer 8, 83–96 (2001).

    Article  CAS  PubMed  Google Scholar 

  35. Bellacosa, A. et al. Molecular alterations of the AKT2 oncogene in ovarian and breast carcinomas. Int. J. Cancer 64, 280–285 (1995).

    Article  CAS  PubMed  Google Scholar 

  36. Simpson, L. & Parsons, R. PTEN: life as a tumor suppressor. Exp. Cell Res. 264, 29–41 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Sebti, S. M. & Hamilton, A. D. Farnesyltransferase and geranylgeranyltransferase I inhibitors and cancer therapy: lessons from mechanism and bench-to-bedside translational studies. Oncogene 19, 6584–6593 (2000).

    Article  CAS  PubMed  Google Scholar 

  38. Cox, A. D. & Der, C. J. Farnesyltransferase inhibitors: promises and realities. Curr. Opin. Pharmacol. 2, 388–393 (2002).

    Article  CAS  PubMed  Google Scholar 

  39. Kohl, N. E. et al. Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice. Nature Med. 1, 792–797 (1995). The study that launched massive interest in farnesyl-transferase inhibitors as tumour therapeutic drugs aimed at RAS. Unfortunately, the activity turned out to be selective for HRAS, an isoform that is only very rarely activated in human tumours.

    Article  CAS  PubMed  Google Scholar 

  40. Lobell, R. B. et al. Evaluation of farnesyl:protein transferase and geranylgeranyl:protein transferase inhibitor combinations in preclinical models. Cancer Res. 61, 8758–8768 (2001).

    CAS  PubMed  Google Scholar 

  41. Prendergast, G. C. Actin' up: RhoB in cancer and apoptosis. Nature Rev. Cancer 1, 162–168 (2001).

    Article  CAS  Google Scholar 

  42. Ohkanda, J., Knowles, D. B., Blaskovich, M. A., Sebti, S. M. & Hamilton, A. D. Inhibitors of protein farnesyltransferase as novel anticancer agents. Curr. Top. Med. Chem. 2, 303–323 (2002).

    Article  CAS  PubMed  Google Scholar 

  43. Singh, S. B. & Lingham, R. B. Current progress on farnesyl protein transferase inhibitors. Curr. Opin. Drug. Discov. Devel. 5, 225–244 (2002).

    CAS  PubMed  Google Scholar 

  44. Cortes, J. E. et al. Efficacy of the farnesyl transferase inhibitor, ZARNESTRATM (R115777), in chronic myeloid leukemia and other other hematological malignancies. Blood 31, 31 Oct 31 2002 [epub ahead of print].

    Google Scholar 

  45. Crooke, S. T. Potential roles of antisense technology in cancer chemotherapy. Oncogene 19, 6651–6659 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Mukhopadhyay, T., Tainsky, M., Cavender, A. C. & Roth, J. A. Specific inhibition of K-ras expression and tumorigenicity of lung cancer cells by antisense RNA. Cancer Res. 51, 1744–1748 (1991).

    CAS  PubMed  Google Scholar 

  47. Chen, G., Oh, S., Monia, B. P. & Stacey, D. W. Antisense oligonucleotides demonstrate a dominant role of c-Ki-RAS proteins in regulating the proliferation of diploid human fibroblasts. J. Biol. Chem. 271, 28259–28265 (1996).

    Article  CAS  PubMed  Google Scholar 

  48. Monia, B. P., Johnston, J. F., Geiger, T., Muller, M. & Fabbro, D. Antitumor activity of a phosphorothioate antisense oligodeoxynucleotide targeted against C-raf kinase. Nature Med. 2, 668–675 (1996).

    Article  CAS  PubMed  Google Scholar 

  49. HŸser, M. et al. MEK kinase activity is not necessary for Raf-1 function. EMBO J. 20, 1940–1951 (2001).

    Article  Google Scholar 

  50. Coudert, B. et al. Phase II trial with ISIS 5132 in patients with small-cell (SCLC) and non-small cell (NSCLC) lung cancer. A European Organization for Research and Treatment of Cancer (EORTC) Early Clinical Studies Group report. Eur. J. Cancer 37, 2194–2198 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Sawyers, C. L. Rational therapeutic intervention in cancer: kinases as drug targets. Curr. Opin. Genet. Dev. 12, 111–115 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Hoshino, R. et al. Constitutive activation of the 41-/43-kDa mitogen-activated protein kinase signaling pathway in human tumors. Oncogene 18, 813–822 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Dudley, D. T., Pang, L., Decker, S. J., Bridges, A. J. & Saltiel, A. R. A synthetic inhibitor of the mitogen-activated protein kinase cascade. Proc. Natl Acad. Sci. USA 92, 7686–7689 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Favata, M. F. et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J. Biol. Chem. 273, 18623–18632 (1998).

    Article  CAS  PubMed  Google Scholar 

  55. Sebolt-Leopold, J. S. Development of anticancer drugs targeting the MAP kinase pathway. Oncogene 19, 6594–6599 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Sebolt-Leopold, J. S. et al. Blockade of the MAP kinase pathway suppresses growth of colon tumors in vivo. Nature Med. 5, 810–816 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Lyons, J. F., Wilhelm, S., Hibner, B. & Bollag, G. Discovery of a novel Raf kinase inhibitor. Endocr. Relat. Cancer. 8, 219–225 (2001).

    Article  CAS  PubMed  Google Scholar 

  58. Davies, H. et al. Mutations of the BRAF gene in human cancer. Nature 417, 949–954 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Normanno, N., Bianco, C., De Luca, A. & Salomon, D. S. The role of EGF-related peptides in tumor growth. Front. Biosci. 6, D685–D707 (2001).

    Article  CAS  PubMed  Google Scholar 

  60. Sibilia, M. et al. The EGF receptor provides an essential survival signal for SOS-dependent skin tumor development. Cell 102, 211–220 (2000). An elegant transgenic mouse study showing that the ability of activated endogenous RAS protein to promote tumour formation is dependent on the activity of EGFR, indicating the importance of autocrine EGF signalling in RAS transformation.

    Article  CAS  PubMed  Google Scholar 

  61. Fabbro, D., Parkinson, D. & Matter, A. Protein tyrosine kinase inhibitors: new treatment modalities? Curr. Opin. Pharmacol. 2, 374–381 (2002).

    Article  CAS  PubMed  Google Scholar 

  62. Wakeling, A. E. Epidermal growth factor receptor tyrosine kinase inhibitors. Curr. Opin. Pharmacol. 2, 382–387 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. de Bono, J. S. & Rowinsky, E. K. The ErbB receptor family: a therapeutic target for cancer. Trends Mol. Med. 8, S19–S26 (2002).

    Article  CAS  PubMed  Google Scholar 

  64. Herbst, R. S. ZD1839: targeting the epidermal growth factor receptor in cancer therapy. Expert Opin. Investig. Drugs 11, 837–849 (2002).

    Article  CAS  PubMed  Google Scholar 

  65. Hidalgo, M. et al. Phase I and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase inhibitor, in patients with advanced solid malignancies. J. Clin. Oncol. 19, 3267–3279 (2001).

    Article  CAS  PubMed  Google Scholar 

  66. Schulze, A., Lehmann, K., Jefferies, H. B., McMahon, M. & Downward, J. Analysis of the transcriptional program induced by Raf in epithelial cells. Genes Dev. 15, 981–994 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Herbst, R. S. & Shin, D. M. Monoclonal antibodies to target epidermal growth factor receptor-positive tumors: a new paradigm for cancer therapy. Cancer 94, 1593–1611 (2002).

    Article  CAS  PubMed  Google Scholar 

  68. Robert, F. et al. Phase I study of anti-epidermal growth factor receptor antibody cetuximab in combination with radiation therapy in patients with advanced head and neck cancer. J. Clin. Oncol. 19, 3234–3243 (2001).

    Article  CAS  PubMed  Google Scholar 

  69. Slamon, D. J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001). The results of the Phase III trials with trastuzumab (Herceptin) that showed it to be an effective treatment for metastatic breast cancer with amplification of ERBB2.

    Article  CAS  PubMed  Google Scholar 

  70. Xie, Y., Li, K. & Hung, M. C. Tyrosine phosphorylation of Shc proteins and formation of Shc/Grb2 complex correlate to the transformation of NIH3T3 cells mediated by the point-mutation activated neu. Oncogene 10, 2409–2413 (1995).

    CAS  PubMed  Google Scholar 

  71. Keshamouni, V. G., Mattingly, R. R. & Reddy, K. B. Mechanism of 17-β-estradiol-induced Erk1/2 activation in breast cancer cells: A role for HER2 and PKC-δ. J. Biol. Chem. 277, 22558–22565 (2002).

    Article  CAS  PubMed  Google Scholar 

  72. Hunger, R. E. et al. Successful induction of immune responses against mutant ras in melanoma patients using intradermal injection of peptides and GM-CSF as adjuvant. Exp. Dermatol. 10, 161–167 (2001).

    Article  CAS  PubMed  Google Scholar 

  73. Reuveni, H. et al. Toward a PKB inhibitor: modification of a selective PKA inhibitor by rational design. Biochemistry 41, 10304–10314 (2002).

    Article  CAS  PubMed  Google Scholar 

  74. Bergo, M. O. et al. Absence of CAAX endoprotease Rce 1: effects on cell growth and transformation. Mol. Cell. Biol. 22, 171–181 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kloog, Y., Cox, A. D. & Sinensky, M. Concepts in Ras-directed therapy. Exp. Opin. Investig. Drugs 8, 2121–2140 (1999).

    Article  CAS  Google Scholar 

  76. Kimoto, M., Sakamoto, K., Shirouzu, M., Mirao, I. & Yokoyama, S. RNA aptamers that specifically bind to the Ras-binding domain of c-Raf-1. FEBS Lett. 441, 322–326 (1998).

    Article  CAS  PubMed  Google Scholar 

  77. Kato-Stankiewicz, J. et al. Inhibitors of Ras/Raf-1 interaction identified by two-hybrid screening revert Ras-dependent transformation phenotypes in human cancer cells. Proc. Natl Acad. Sci. USA 99, 14398–14403 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ahmadian, M. R. et al. Guanosine triphopshatase stimulation of oncogenic Ras mutants. Proc. Natl Acad. Sci. USA 96, 7065–7070 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Hidalgo, M. & Rowinsky, E. K. The rapamycin-sensitive signal transduction pathway as a target for cancer therapy. Oncogene 19, 6680–6686 (2000).

    Article  CAS  PubMed  Google Scholar 

  80. Neckers, L. Hsp90 inhibitors as novel cancer chemotherapeutic agents. Trends Mol. Med. 8, S55–S61 (2002).

    Article  CAS  PubMed  Google Scholar 

  81. Zucker, S., Cao, J. & Chen, W. T. Critical appraisal of the use of matrix metalloproteinase inhibitors in cancer treatment. Oncogene 19, 6642–6650 (2000).

    Article  CAS  PubMed  Google Scholar 

  82. Rosen, L. S. Angiogenesis inhibition in solid tumors. Cancer J. 7 (Suppl. 3), S120–S128 (2001).

    PubMed  Google Scholar 

  83. Kirn, D., Niculescu-Duvaz, I., Hallden, G. & Springer, C. J. The emerging fields of suicide gene therapy and virotherapy. Trends Mol. Med. 8, S68–S73 (2002).

    Article  CAS  PubMed  Google Scholar 

  84. Biederer, C., Ries, S., Brandts, C. H. & McCormick, F. Replication-selective viruses for cancer therapy. J. Mol. Med. 80, 163–175 (2002).

    Article  CAS  PubMed  Google Scholar 

  85. Raff, M. C. Social controls on cell survival and cell death. Nature 356, 397–400 (1992).

    Article  CAS  PubMed  Google Scholar 

  86. End, D. W. et al. Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Res. 61, 131–137 (2001).

    CAS  PubMed  Google Scholar 

  87. Rose, W. C. et al. Preclinical antitumor activity of BMS-214662, a highly apoptotic and novel farnesyltransferase inhibitor. Cancer Res. 61, 7507–7517 (2001). References 86 and 87 describe preclinical data on two of the leading farnesyltransferase inhibitors. The significant effects that are seen in these model systems are probably not due to targeting RAS, and it remains unclear whether they can be replicated in the clinic.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASES

Cancer.gov

breast carcinoma

colon carcinoma

colorectal cancer

head and neck cancer

leukaemia

melanoma

non-small-cell lung carcinoma

ovarian carcinoma

prostate carcinoma

renal-cell carcinoma

stomach carcinoma

LocusLink

ABL

AKT

BCR

CDC42

EGFR

ELK1

ERBB2

FOS

FoxO family

GRB2

HRAS

HSP90

ICMT

JUN

KRAS

MAPKs

MEK1

MEK2

NF1

NRAS

PDK1

PTEN

RAC

RAF

RALBP1

RALGDS

RGL

RB

RGL2

RHO

SHC

SOS1

SOS2

TP53

TGF-α

VEGF

Glossary

SH2 DOMAIN

Src homology 2 domain. A protein domain capable of binding tyrosine phosphorylated sites.

SH3 DOMAIN

Src homology 3 domain. A protein domain capable of binding proline-rich motifs.

AKT/PKB

A family of three closely related serine/threonine protein kinases containing amino-terminal pleckstrin homology domains that bind to the lipid products of phosphatidylinositol 3-kinase (PI3K). They are activated on PI3K stimulation and provide anti-apoptotic signals to the cell.

FORKHEAD

A large superfamily of transcription factors, of which one family, FoxO, is phosphorylated and inhibited by AKT/PKB.

BIM

A member of the BCL-2 family of apoptosis regulatory proteins. BIM induces apoptosis by acting at mitochondria to promote the release of cytochrome c into the cytosol.

FAS LIGAND

An extracellular protein that binds and activates the death receptor FAS, also known as CD95 and APO1, leading to initiation of apoptosis.

p110α

One isoform of the catalytic subunit of phosphoinositide 3-kinase (PI3K). Type I PI3Ks are made up of either α, β, γ or δ forms of p110, together with a regulatory p85 subunit (for p110α, β and δ) or p101 subunit (for p110γ).

IC50

The concentration of a drug giving a 50% inhibition of the activity of a target enzyme.

RNA APTAMER

A synthetic RNA molecule selected for ability to interact with a target protein.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer 3, 11–22 (2003). https://doi.org/10.1038/nrc969

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc969

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing